Pharmaceutical compositions can include ceftolozane lyophilized in the absence of tazobactam.

Patent
   8968753
Priority
Mar 15 2013
Filed
May 22 2014
Issued
Mar 03 2015
Expiry
Mar 14 2034

TERM.DISCL.
Assg.orig
Entity
Large
7
351
currently ok
1. A method of treating an infection selected from the group consisting of a complicated intra-abdominal infection and a complicated urinary tract infection in a patient comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising (a) ceftolozane lyophilized in the absence of tazobactam and (b) a tazobactam composition, in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active.
9. A method of treating an infection in a patient selected from the group consisting of a complicated intra-abdominal infection and a complicated urinary tract infection, the method comprising intravenously administering to the patient a therapeutically effective amount of ceftolozane obtained by lyophilizing a solution comprising ceftolozane without tazobactam to obtain a ceftolozane composition, and tazobactam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active.
28. A method of treating an infection selected from the group consisting of a complicated intra-abdominal infection and a complicated urinary tract infection in a patient comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising:
(a) tazobactam or a pharmaceutically acceptable salt thereof and
(b) a lyophilized ceftolozane composition, wherein prior to lyophilization the ceftolozane composition does not contain tazobactam or salts thereof.
18. A method of treating an infection selected from the group consisting of a complicated intra-abdominal infection and a complicated urinary tract infection in a patient comprising:
dissolving in a pharmaceutically acceptable carrier a composition comprising tazobactam and ceftolozane prepared in the absence of tazobactam to obtain a pharmaceutical composition comprising ceftolozane active and tazobactam active in a 2:1 weight ratio; and
administering to the patient a therapeutically effective amount of the pharmaceutical composition.
2. The method of claim 1, wherein the infection is a complicated urinary tract infection.
3. The method of claim 1, wherein the pharmaceutical composition is intravenously administered to the patient as an infusion after combining the ceftolozane lyophilized in the absence of tazobactam and the tazobactam composition with a pharmaceutically acceptable carrier.
4. The method of claim 1, wherein the pharmaceutical composition comprises an amount of a pharmaceutically acceptable salt of ceftolozane providing a total of 1,000 mg of ceftolozane active, blended with the tazobactam composition to form the pharmaceutical composition.
5. The method of claim 1, wherein the tazobactam in the tazobactam composition is tazobactam sodium.
6. The method of claim 1, wherein the tazobactam in the tazobactam composition is a crystalline tazobactam.
7. The method of claim 1, wherein the ceftolozane is ceftolozane sulfate.
8. The method of claim 1, wherein the pharmaceutical composition does not contain a compound of formula (III)
##STR00006##
10. The method of claim 9, wherein the ceftolozane and the tazobactam are administered in a pharmaceutical composition comprising an amount of a pharmaceutically acceptable salt of ceftolozane providing a total of 1,000 mg of ceftolozane active.
11. The method of claim 10, wherein the pharmaceutical composition is obtained by a process comprising the step of combining tazobactam lyophilized in the absence of ceftolozane with the lyophilized ceftolozane composition.
12. The method of claim 9, wherein the tazobactam is tazobactam sodium.
13. The method of claim 9, wherein the tazobactam is a crystalline tazobactam.
14. The method of claim 9, wherein the ceftolozane is ceftolozane sulfate.
15. The method of claim 10, wherein the pharmaceutical composition does not contain a compound of formula (III)
##STR00007##
16. The method of claim 10, wherein the ceftolozane and the tazobactam are lyophilized in separate solutions prior to forming the pharmaceutical composition.
17. The method of claim 9, wherein the ceftolozane composition is obtained by dissolving a compound of formula (I):
##STR00008##
in the solution, adjusting the pH of the solution to about 5-7 prior to lyophilization, and lyophilizing the solution to obtain the lyophilized ceftolozane composition.
19. The method of claim 18, wherein the infection is a complicated urinary tract infection.
20. The method of claim 18, wherein the pharmaceutical composition is intravenously administered to the patient as an infusion.
21. The method of claim 20, wherein the pharmaceutical composition comprises an amount of a pharmaceutically acceptable salt of ceftolozane providing a total of 1,000 mg of ceftolozane active.
22. The method of claim 18, wherein the ceftolozane is obtained by a process comprising the step of lyophilizing a solution comprising ceftolozane sulfate in the absence of tazobactam.
23. The method of claim 20, wherein the pharmaceutical composition is obtained by a process comprising the step of combining tazobactam with the lyophilized ceftolozane composition.
24. The method of claim 18, wherein the tazobactam is tazobactam sodium.
25. The method of claim 18, wherein the pharmaceutical composition does not contain a compound of formula (III)
##STR00009##
26. The method of claim 18, wherein the ceftolozane and the tazobactam are lyophilized in separate solutions prior to administering to the patient.
27. The method of claim 18, wherein the ceftolozane is lyophilized in the absence of tazobactam and is prepared by lyophilizing a solution of ceftolozane of formula (I) in the absence of tazobactam, adjusted to a pH to about 5-7 prior to lyophilization:
##STR00010##
29. The method of claim 28, wherein the pharmaceutical composition comprises the ceftolozane and the tazobactam in a weight ratio of about 2:1 between the ceftolozane active and the tazobactam active.
30. The method of claim 29, wherein the lyophilized ceftolozane is lyophilized in a solution comprising ceftolozane of formula (I) in the absence of tazobactam:
##STR00011##
adjusted to a pH of about 5-7 prior to lyophilization.

This application is a continuation of U.S. patent application Ser. No. 14/214,212, filed Mar. 14, 2014, which claims priority to U.S. Provisional Patent Application No. 61/792,092, filed Mar. 15, 2013, and U.S. Provisional Patent Application No. 61/793,007, filed Mar. 15, 2013, both of which are incorporated herein in their entirety.

This disclosure relates to antibacterial compositions comprising ceftolozane and tazobactam.

The pharmaceutical antibiotic composition comprising ceftolozane and tazobactam in a 2:1 weight ratio of ceftolozane active to tazobactam active (“CXA-201”) displays potent antibacterial activity, including antibiotic activity against infections caused by many Gram-negative pathogens such as Pseudomonas aeruginosa (P. aeruginosa), Escherichia coli (E. coli), Klebsiella pneumonia (K. pneumonia). In particular, CXA-201 is a pharmaceutical composition useful for intravenous administration for the treatment of complicated intra-abdominal infections and/or complicated urinary tract infections, and is being evaluated for treatment of pneumonia.

Ceftolozane is a cephalosporin antibacterial agent, also referred to as CXA-101, FR264205, or by chemical names such as (6R,7R)-3-[(5-amino-4-{[(2-aminoethyl)carbamoyl]amino}-1-methyl-1H-pyrazol-2-ium-2-yl)methyl]-7-({(2Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-[(1-carboxy-1-methylethoxy)imino]acetyl}amino)-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate, and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate. The antibacterial activity of ceftolozane is believed to result from its interaction with penicillin binding proteins (PBPs) to inhibit the biosynthesis of the bacterial cell wall which acts to stop bacterial replication. Ceftolozane sulfate is a pharmaceutically acceptable ceftolozane salt of formula (I) that can be formulated for intravenous administration or infusion.

##STR00001##

In CXA-201, ceftolozane is combined with the β-lactamase inhibitor (“BLI”) tazobactam. Tazobactam is a BLI against Class A and some Class C β-lactamases, with well-established in vitro and in vivo efficacy in combination with active β-lactam antibiotics. Tazobactam can be combined with ceftolozane as a free acid tazobactam form of formula (II).

##STR00002##

Pharmaceutical compositions comprising one or more drug substances can be prepared by lyophilization of a solution containing the drug substance(s). Lyophilization is a process of freeze-drying in which water is sublimed from a frozen solution of one or more solutes. Specific methods of lyophilization are described in Remington's Pharmaceutical Sciences, Chapter 84, page 1565, Eighteenth Edition, A. R. Gennaro, (Mack Publishing Co., Easton, Pa., 1990). It has now been found and reported herein that compositions formed by lyophilizing ceftolozane and tazobactam through co-lyophilization, (i.e., the ceftolozane and tazobactam were combined and lyophilized together in Example 3, as opposed to separately) resulted in the formation of significant amounts of an undesired by-product represented by formula (III) (See Example 5 and the results reported in Tables 5 and 6 in FIGS. 7A and 7B).

##STR00003##

Therefore, there remains a need for compositions comprising ceftolozane and/or tazobactam with reduced or even undectable amounts (e.g., less than about 0.03% by high performance liquid chromatography, or “HPLC”) of the compound of formula (III) and methods for manufacturing these compositions.

Applicants have discovered pharmaceutical compositions comprising ceftolozane and tazobactam with reduced and even undetectable amounts of the compound of Formula (III), and methods of manufacturing these compositions. The invention is based in part on the discovery that the formation of the compound represented by Formula (III) can be reduced if not completely suppressed by lyophilizing ceftolozane in the absence of tazobactam and then blending the lyophilized ceftolozane with a dry tazobactam composition, such as a tazobactam composition lyophilized in the absence of ceftolozane (See Example 6 and the results reported in Tables 8 and 9 in FIGS. 9A and 9B). Based on these results, pharmaceutical compositions comprising ceftolozane and tazobactam, and pharmaceutical compositions prepared using ceftolozane and tazobactam are provided herein. In particular, these pharmaceutical compositions can include ceftolozane and/or tazobactam with reduced or even undetectable amounts of a compound of formula (III).

##STR00004##

In one embodiment, a pharmaceutical composition can include ceftolozane and tazobactam with less than 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC or even undectable amounts of the compound of formula (III) (e.g., less than about 0.03% of the compound of Formula (III) measured by HPLC). These pharmaceutical compositions can be obtained by a process comprising the steps of (a) lyophilizing ceftolozane in the absence of tazobactam to obtain a lyophilized ceftolozane composition; and (b) combining the lyophilized ceftolozane with tazobactam under conditions suitable to obtain said pharmaceutical composition with the aforementioned purity levels. The combination of the lyophilized ceftolozane composition with tazobactam can include blending the lyophilized ceftolozane composition with lyophilized or crystalline tazobactam material.

In one aspect, provided herein is a pharmaceutical composition comprising a blend of separately lyophilized tazobactam and ceftolozane sulfate in an amount providing 1,000 mg of ceftolozane active per 500 mg of tazobactam active, further comprising less than 0.15%, 0.10%, 0.05% or 0.03% by weight; from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC; or even undectable amounts (e.g., less than about 0.03% by HPLC) of a compound of formula (III) detectable at a retention time relative to ceftolozane of 1.22 by high performance liquid chromatography (HPLC) using a Develosil column ODS-UG-5; 5 micrometers; 250×4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45° C. (hereinafter referred to as the “method of Example 2”).

CXA-201 compositions comprising less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC of the compound of formula (III) can be obtained by a process comprising the steps of: (a) forming a first aqueous solution comprising ceftolozane (e.g., in a pharmaceutically acceptable salt such as formula (I)), (b) lyophilizing the first aqueous solution to obtain a lyophilized ceftolozane composition, and (c) blending the lyophilized ceftolozane composition with a tazobactam composition (e.g., tazobactam acid lyophilized in the absence of ceftolozane) in an amount that provides a 2:1 weight ratio between the amount of ceftolozane active and tazobactam active.

FIGS. 1A and 1B are chromatograms of CXA-101 ceftolozane drug substance obtained from the lyophilization process of Example 1. The chromatograms were obtained according to the analytical method described in Example 2.

FIG. 2 is a diagram of a lyophilization process for the ceftolozane obtained according to the process described in Example 1.

FIG. 3 is a table (Table 1) of peaks for the ceftolozane prepared by the lyophilization process in Example 1 obtained by HPLC according to the analytical method of Example 2.

FIG. 4A is a schematic showing a process for making the compound of formula (III) with ceftolozane and tazobactam.

FIG. 4B is a table (Table 2) showing a first composition that can be lyophilized to form a composition comprising the compound of formula (III).

FIG. 5 is a table (Table 3) showing in-process testing of blending samples of bulk drug product at five places.

FIG. 6 is a table (Table 4) of the composition of the formulation used to prepared the Co-Lyophilized Combo drug product used in Example 5.

FIG. 7A is a table (Table 5) of the impurity composition of the Co-Lyophilized Combo drug product at T0 (time zero), T1 (one month) and T2 (three months) after being maintained at 25° C./60% relative humidity.

FIG. 7B is a table (Table 6) of the impurity composition of the Co-Lyophilized Combo drug product at T0 (time zero), T1 (one month) and T2 (three months) after being maintained at 40° C./75% relative humidity.

FIG. 8 is a table (Table 7) of the composition of the formulation used to prepare the Blended Combination drug product used in Example 6.

FIG. 9A is a table (Table 8) of the impurity composition of the Blended Combination drug product at T0 (time zero), T1 (one month) and T2 (three months) after being maintained at 25° C./60% relative humidity.

FIG. 9B is a table (Table 9) of the impurity composition of the Blended Combination drug product at T0 (time zero), T1 (one month) and T2 (three months) after being maintained at 40° C./75% relative humidity.

Pharmaceutical compositions comprising ceftolozane and tazobactam with reduced or even undectable levels of the compound of formula (III) (e.g., including levels of compound of formula (III) that are not detectable by HPLC according to Example 2 and/or comprise less than 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC according to Example 2) can be obtained by blending a first composition comprising a therapeutically effective amount of ceftolozane in the absence of tazobactam with a second composition comprising a therapeutically effective amount of tazobactam in the absence of ceftolozane to form a blended pharmaceutical composition.

The (first) ceftolozane composition can be prepared in the absence of tazobactam by forming a first aqueous solution comprising ceftolozane sulfate and other components including excipients, stabilizers, pH adjusting additives (e.g., buffers) and the like. Non-limiting examples of these additives include sodium chloride, citric acid and L-arginine. For example, the use of sodium chloride results in greater stability; L-arginine is used to adjust the aqueous solution to a pH of 5-7 (e.g., to pH 6-7) and to increase the solubility of ceftolozane; and citric acid is used to prevent discoloration of the product, due to its ability to chelate metal ions. In one embodiment, the pH of the first aqueous solution is suitable for making an injectable product (e.g,, a pH range of 5-7, including 6-7). Preferably, the first aqueous solution comprises about 125 mg-500 mg sodium chloride per 1,000 mg of ceftolozane active. The ceftolozane can be included as an amount of ceftolozane sulfate of formula (I) containing at least about 1,000 mg ceftolozane active. The (first) aqueous solution is then lyophilized to form a first lyophilized ceftolozane composition, which is combined with tazobactam, e.g., the lyophilized tazobactam (e.g., lyophilized tazobactam sodium) or crystalline tazobactam

The (second) tazobactam composition can be prepared in the absence of ceftolozane by forming a second solution comprising tazobactam. The tazobactam can be included in an amount providing about 500 mg of tazobactam active per 1,000 mg ceftolozane active (i.e., a 1:2 weight ratio of tazobactam active to ceftolozane active). Tazobactam is a β-lactamase inhibitor of the structure of formula (II) in its free acid form.

##STR00005##

Unless otherwise indicated, tazobactam can be a free acid, a sodium salt, an arginine salt, or a hydrate or solvate thereof. In one embodiment, the tazobactam in the (second) tazobactam composition is tazobactam acid and the second composition further comprises sodium bicarbonate or sodium hydroxide. Lyophilizing tazobactam in the presence of sodium bicarbonate or sodium hydroxide forms a lyophilized tazobactam sodium, which can then be further blended with the (first) lyophilized ceftolozane composition.

Pharmaceutical compositions with reduced or undectable amounts of the compound of formula (III) can be obtained by lyophilizing ceftolozane without formylacetic acid and/or tazobactam under conditions that prevent formation of the compound of formula (III) (e.g., Example 1 and 4). The presence of a compound of formula (III) can be detected by HPLC (e.g., Examples 2, 5 and 6). Specific methods of lyophilization are described in Example 1 and Remington's Pharmaceutical Sciences, Chapter 84, page 1565, Eighteenth Edition, A. R. Gennaro, (Mack Publishing Co., Easton, Pa., 1990). The formation of the compound of formula (III) can be avoided by preventing the reaction of ceftolozane and formylacetic acid. In one embodiment, the compound of formula (III) can be suppressed by separately lyophlizing ceftolozane sulfate and tazobactam in separate solutions, and then blending the lyophilized compositions to form a pharmaceutical composition.

In one aspect, antibiotic pharmaceutical compositions comprising ceftolozane and tazobactam with less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC of the compound of formula (III) are obtained by a process comprising the steps of: (a) lyophilizing ceftolozane in the absence of tazobactam to obtain a lyophilized ceftolozane composition, and (b) blending the lyophilized ceftolozane composition with a composition comprising tazobactam under conditions suitable for attaining the aforementioned purity levels, e.g., by blending with crystalline tazobactam or lyophilized tazobactam.

In another aspect, antibiotic pharmaceutical compositions comprising ceftolozane and tazobactam and less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC of the compound of formula (III) are obtained by a process comprising the steps of: (a) lyophilizing tazobactam in the absence of ceftolozane to obtain a lyophilized tazobactam composition, and (b) blending the lyophilized tazobactam composition with a composition comprising ceftolozane (e.g., lyophilized ceftolozane sulfate).

In a third aspect, antibiotic pharmaceutical compositions comprising ceftolozane and tazobactam and less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC of the compound of formula (III) are obtained by a process comprising the steps of: (a) lyophilizing tazobactam in the absence of ceftolozane to obtain a lyophilized tazobactam composition, (b) lyophilizing ceftolozane in the absence of tazobactam to obtain a lyophilized ceftolozane composition, and (c) blending the lyophilized tazobactam composition with the lyophilized ceftolozane composition.

Pharmaceutical compositions comprising the compound of formula (III), ceftolozane and tazobactam can be formulated to treat infections by parenteral administration (including subcutaneous, intramuscular, and intravenous) administration. Pharmaceutical compositions may additionally comprise excipients, stabilizers, pH adjusting additives (e.g., buffers) and the like. Non-limiting examples of these additives include sodium chloride, citric acid and L-arginine. For example, the use of sodium chloride results in greater stability; L-arginine is used to adjust pH and to increase the solubility of ceftolozane; and citric acid is used to prevent discoloration of the product, due to its ability to chelate metal ions. In one particular embodiment, the pharmaceutical compositions described herein are formulated for administration by intravenous injection or infusion.

Other pharmaceutical antibiotic compositions can include ceftolozane sulfate and the compound of formula (III). For example, pharmaceutical compositions comprising 0.13%, 0.15%, 0.30%, 0.38%, 0.74% or 0.97% of the compound of formula (III) are herein. The pharmaceutical antibiotic compositions can be provided in a unit dosage form (e.g., in a vial). The unit dosage form can be dissolved with a pharmaceutically acceptable carrier, and then intravenously administered. The unit dosage form comprises 1000 mg of ceftolozane active and 500 mg tazobactam, typically 1000 mg ceftolozane active as ceftolozane sulfate and 500 mg of tazobactam active as tazobactam sodium, argininate or free acid. The unit dosage forms are commonly stored in vials.

In one aspect, provided herein is a unit dosage form container (e.g., a bag, vial or the like) containing a unit dosage form of a pharmaceutical composition formulated for parenteral administration for the treatment of complicated intra-abdominal infections or complicated urinary tract infections, the pharmaceutical composition comprising a therapeutically effective amount of ceftolozane sulfate and tazobactam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active, the pharmaceutical composition obtained by a process comprising the steps of:

In one embodiment of the unit dosage form container, the tazobactam in the second solution is tazobactam acid, and wherein the tazobactam acid in the second solution is lyophilized in the presence of sodium bicarbonate or sodium hydroxide, thereby forming lyophilized tazobactam sodium in the second lyophilized tazobactam solution. A pharmaceutical composition can include ceftolozane sulfate and tazobactam in an amount providing 1,000 mg of ceftolozane active per 500 mg of tazobactam active, and 0.03% to 0.15% by HPLC of a compound of formula (III) detectable at a retention time relative to ceftolozane of 1.22 by high performance liquid chromatography using a Develosil column ODS-UG-5; 5 micrometers; 250×4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45° C. Optionally, the pharmaceutical composition can further include 125 mg to 500 mg of sodium chloride per 1,000 mg of ceftolozane active, and L-arginine. The tazobactam in the composition can be tazobactam sodium.

The pharmaceutical compositions provided herein comprising ceftolozane sulfate and tazobactam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active, can be obtained by a process comprising the steps of:

The pharmaceutical compositions can be administered for the treatment of infections, such as complicated intra-abdominal infections, complicated urinary tract infections (cUTIs) and pneumonia (e.g., community-acquired, hospital-acquired, etc). In one aspect, provided herein is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition prepared according to the methods described herein. A method for the treatment of bacterial infections in a mammal can comprise administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane sulfate and sodium chloride.

Non-limiting examples of bacterial infections that can be treated by the methods of the invention include infections caused by: aerobic and facultative gram-positive microorganisms (e.g., Staphylococcus aureus, Enterococcus faecalis, Staphylococcus epidermidis, Streptococcus agalactiae, Streptococcus pneumonia, Streptococcus pyogenes, Viridans group streptococci), aerobic and facultative gram-negative microorganisms (e.g., Acinetobacter baumanii, Escherichia coli, Haemophilus influenza, Klebsiella pneumonia, Pseudomonas aeruginosa, Citrobacter koseri, Moraxella catarrhalis, Morganella morganii, Neisseria gonorrhoeae, Proteus mirabilis, Proteus vulgaris, Serratia marcescens, Providencia stuartii, Providencia rettgeri, Salmonella enterica), gram-positive anaerobes (Clostridium perfringens), and gram-negative anaerobes (e.g., Bacteroides fragilis group (e.g., B. fragilis, B. ovatus, B. thetaiotaomicron, and B. vulgates), Bacteroides distasonis, Prevotella melaninogenica).

In certain embodiments of the methods described herein, bacterial infection is associated with one or more of the following conditions: complicated intra-abdominal infections, complicated urinary tract infections (cUTIs) and pneumonia (e.g., community-acquired, or nosocomial pneumonia). Community-acquired pneumonia (moderate severity only) can include infections caused by piperacillin-resistant, beta-lactamase producing strains of Haemophilus influenza. Nosocomial pneumonia (moderate to severe) caused by piperacillin-resistant, beta-lactamase producing strains of Staphylococcus aureus and by Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumoniae, and Pseudomonas aeruginosa.

As used herein, “treating”, “treat” or “treatment” describes the management and care of a patient for the purpose of combating a disease, condition, or disorder and includes the administration of a pharmaceutical composition of the present invention to alleviate the symptoms or complications of a disease, condition or disorder, or to reduce the extent of the disease, condition or disorder. The term “treat” can also include treatment of a cell in vitro or an animal model.

By a “therapeutically effective amount” of a compound of the invention is meant a sufficient amount of the compound to treat the disorder (e.g., bacterial infection). The specific therapeutically effective amount that is required for the treatment of any particular patient or organism (e.g., a mammal) will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound or composition employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts (see, for example, Goodman and Gilman's, “The Pharmacological Basis of Therapeutics”, Tenth Edition, A. Gilman, J. Hardman and L. Limbird, eds., McGraw-Hill Press, 155-173, 2001, which is incorporated herein by reference in its entirety). The therapeutically effective amount for a given situation can be readily determined by routine experimentation and is within the skill and judgment of the ordinary clinician.

As used herein, the term “ceftolozane active” refers to active portion of a salt form of ceftolozane in the free base form of ceftolozane.

As used herein, the term “tazobactam active” refers to the active portion of a salt form of tazobactam in the tazobactam free acid form.

As used herein, “1,000 mg of ceftolozane as ceftolozane active” refers to an amount of ceftolozane sulfate effective to provide 1,000 mg of ceftolozane active. The amount of sodium chloride per gram of ceftolozane activity in a pharmaceutical composition containing ceftolozane sulfate and sodium chloride can be calculated using the relevant molecular weights of ceftolozane, ceftolozane sulfate, sodium chloride and sodium. As used herein, “500 mg of tazobactam as tazobactam active” refers to an amount of tazobactam sodium or tazbactam arginine effective to provide 500 mg of tazobactam active.

As used herein, references to an amount of a substance as “% of the compound of . . . ” or “% by HPLC” (unless otherwise indicated) refer to the % of a compound detected by high performance liquid chromatography (HPLC) according to the method of Example 2.

Illustrative Examples of Selected Embodiments of the Invention

There are four main steps in the manufacture of CXA-101 bulk drug product: dissolution, sterile filtration, bulk lyophilization, and packaging into Sterbags®. These four main steps are composed of a total of 20 minor steps. The CXA-101 bulk drug product manufacturing process is presented below.

I. Dissolution

1. The prescribed amount of water for injection (“WFI”) is charged into the dissolution reactor.

2. A prescribed amount of citric acid is added.

3. The solution is cooled at 5° C. to 10° C.

4. A prescribed amount of CXA-101 drug substance is added to the solution.

5. A prescribed amount of L-arginine is slowly added to the solution.

6. A check for complete dissolution is performed. Solution pH is verified to be in the target range of 6.5 to 7.0.

7. A prescribed amount of sodium chloride is added to the solution.

8. A check for complete dissolution is performed. Solution pH is verified to be in the target range of 6.0 to 7.0. If the pH is out of this range adjust with either L-Arginine or citric acid.

9. WFI is added to bring the net weight to 124.4 kg and the solution is mixed well.

10. Samples are withdrawn for testing of final pH.

II. Sterile Filtration

11. The solution is passed through the filter (pore size 0.45 μm) followed by double filters (pore size 0.22 μm) onto a shelf on the Criofarma lyophilizer.

12. The line is washed with WFI.

13. The washing solution is passed from Step 12 through sterile filtration.

III. Bulk Lyophilization

14. The washing solution is loaded onto a separate shelf in the lyophilizer (and later discarded).

15. The solution is lyophilized until dry.

16. The product shelf is cooled to 20° C.±5° C.

IV. Packaging into Sterbags®

17. The lyophilized bulk drug product powder is milled.

18. The milled powder is sieved.

19. The sieved powder is blended for 30 minutes.

20. The powder is then discharged into Sterbags®

Prefiltration and Sterile-Filtration

Filtrate the compounded solution with a sterile tilter-set which consists of a 0.2 um polyvinylidene fluoride membrane filter (Durapore®, Millipore) and a 0.1 urn polyvinylidene fluoride membrane filter (Durapore®, Millipore) connected in tandem. Confirm the integrity of each filter before and after the filtration. Take approximately 100 mL of the filtrate in order to check bioburden.

Filter the prefiltered compounded solution through a sterile filter-set which consists of a 0.2 um polyvinylidene fluoride membrane filter and a 0.1 urn polyvinylidene fluoride membrane filter connected in tandem, and introduce the final filtrate into an aseptic room. Confirm the integrity of each filter before and after the filtration.

Processing of Vial, Stopper and Flip-Off Cap

Wash a sufficient quantity of 28 mL vials with water for injection and sterilize the washed vials by a dry-heat sterilizer. Then transfer the sterilized vials into a Grade A area located in an aseptic room.

Wash a sufficient quantity of stoppers with, water for injection. Sterilize and dry the washed stoppers by steam sterilizer. Then transfer the sterilized stoppers into a Grade A area located in an aseptic room.

Sterilize a sufficient quantity of flip-off caps by steam sterilizer. Then transfer the sterilized flip-off caps into a Grade A or B area located in an aseptic room.

Filling and Partially Stoppering

Adjust the fill weight of the filtered compounded solution to 11.37 g (corresponds to 10 mL of the compounded solution), then start filling operation. Check the filled weight in sufficient frequency and confirm it is in target range (11.37 g±1%, 11.26 to 11.43 g). When deviation from the control range (11.37 g±2%, 11.14 to 11.59 g) is occurred, re-adjust the filling weight.

Immediately after a vial is filled, partially stopper the vial with a sterilized stopper. Load the filled and partially stoppered vials onto the shelves of a lyophilizer aseptically.

Lyophilization to Crimping, Visual Inspection, Labeling and Packaging

After all filled and partially stoppered vials are loaded into a lyophilizer, start the lyophilization program shown in FIG. 2. Freeze the loaded vials at −40° C. and keep until all vials freeze. Forward the program to primary drying step (shelf temperature; −20° C., chamber pressure; 100 to 150 mTorr). Primary drying time should be determined by monitoring the product temperature. Forward the program to secondary drying step (shelf temperature; 30° C., chamber pressure; not more than 10 mTorr) after completion of the primary drying step. After all vials are dried completely, return the chamber pressure to atmospheric pressure with sterilized nitrogen. Then stopper vials completely.

Unload the lyophilized vials from the chamber and crimp with sterilized flip-off caps.

Subject all crimped vials to visual inspection and label and package all passed vials.

A. Operative Conditions

Column Develosil ODS-UG-5; 5 μm, 250 × 4.6 mm (Nomura
Chemical, Japan)
Mobile phase Sodium Perchlorate Buffer Solution (PH 2.5)/CH3CN
90:10 (vlv)
Flow rate 1.0 mL/min
Wavelength 254 nm
Injection volume 10 μL
Oven 45° C.
Temperature
Run Time 85 minutes
Time (min) A % B %
Gradient Profile: 0 75 25
30 70 30
60 0 100
85 0 100
85.1 75 25
110 75 25

B. Mobile Phase Preparation.

Sodium Perchlorate Buffer Solution was made by dissolving 14.05 g of sodium perchlorate Monohydrate in 1000.0 mL of water followed by adjusting pH to 2.5 with diluted perchloric acid (1 in 20).

Mobile Phase was then made by mixing Sodium Perchlorate Buffer Solution (pH 2.5) and acetonitrile in the ratio 90:10 (v/v).

Sodium Acetate Buffer Solution pH 5.5 (Diluent) was made by dissolving 1.36 g of sodium acetate trihydrate in 1000.0 mL of water followed by adjusting to pH 5.5 with diluted acetic acid (1 in 10).

C. Sample Preparation.

Sample solution: dissolve 20.0 mg, exactly weighed, of Sample, in 20.0 mL of water (Prepare just before injection into HPLC system).

System Suitability Solution (1%): take 1.0 mL of the Sample Solution (use first sample if more are present) and transfer into a 100.0 mL volumetric flask, dilute with water to volume and mix.

D. HPLC Analysis Procedure

C i = A i × 100 A t + A i

wherein:

Ci=Amount of related substance i in the Sample, area %

Ai=Peak area of related substance i in the Sample chromatogram

At=Area of CXA-101 peak in the Sample chromatogram

At+Σ Ai=Total peaks area in the Sample chromatogram

Consider as any Unspecified Impurity, each peak in the chromatogram except CXA-101, peaks from 1 to 11 and every peak present in the blank chromatogram and report the largest.

C T = A i × 100 A t + A i

wherein:

CT=total impurities content in the Sample, area %

At=area of CXA-101 peak in the sample chromatogram

Σ Ai=total peak areas of impurities in the sample chromatogram

Compositions comprising the compound of formula (III) were prepared by the process shown in FIG. 4A by (a) forming an aqueous solution comprising the components in Table 2 (FIG. 4B), and (b) lyophilizing the aqueous solution. Sodium content was approximately 78 mg/g of tazobactam in drug product after lyophilization. Water was removed during the lyophilization process and is controlled at no more than 4% by HPLC.

Sterile Dry Blending of Bulk Lyophilized Ceftolozane and Bulk Lyophilized Tazobactam

The CXA-101 produced by Example 1 is blended with lyophilized tazobactam. A low energy drum blender that agitates the material by tumbling and also moving the bed up and down is used. A representative process of blending is described according to the table in FIG. 5. The table in FIG. 5 shows in-process testing of blending samples of bulk drug product at five places. For CXA-101/tazobactam for injection, the blender was charged with 23.4 kg of CXA-101 bulk product, and 5.4 kg of tazobactam bulk product. Both the CXA-101 and tazobactam were individually lyophilized beforehand. The material was blended for 180 minutes. In-process tests of content assay for both CXA-101 and tazobactam were performed to assess the homogeneity using the samples of blend materials taken from three places. The RSD for each of CXA-101 and tazobactam content assay was no greater than 2% and the RSD for the ratio of CXA-101/tazobactam was no greater than 2.2%. (See Table 3 in FIG. 5).

The Co-Lyophilized Combo Drug Product was prepared as described above in Example 3. The formulation composition of the Co-Lyophilized Combo drug product is shown in FIG. 6 (Table 4). This sample was maintained at 25° C./RH=60% and 40° C./RH=75% after one month (T1) and three months (T2). Samples were analyzed using a HPLC method as described in Example 2. The data for analysis of the samples by HPLC is shown in the tables in FIG. 7A (Table 5: Stability data of Co-Lyo Combo Drug Product at 25° C.) and FIG. 7B (Table 6: Stability data Co-Lyo Combo Drug Product at 40° C.). The presence of the compound of Formula (III) was identified has having a retention time of about 1.22 as measured by HPLC (see Example 2). RRT=1.22 was observed in co-lyophilized drug product. The compound of formula (III) is believed to be formed by a reaction between ceftolozane and formylacetic acid, which was a degradation product of tazobactam. The amount of the compound of formula (III) in a composition comprising ceftolozane and tazobactam can be increased over time at 25° C. and at 40° C.

A. Preparation of Blend Combination Drug Product

The blend drug product was prepared, as described above in Example 4, on lab scale by use of a small blender. The composition of the blend drug product is shown in Table 7, FIG. 8. The CXA-101 was obtained by lyophilization of ceftolozane sulfate in the absence of tazobactam, and the tazobactam sodium material was obtained by lyophilization of tazobactam prior to blending of the ceftolozane and tazobactam components.

B. Stress Test

This sample was put into stability study. The following Tables 8 (FIG. 9A) and 9 (FIG. 9B) are representative examples that summarizes the results at 25° C./RH=60% and 40° C./RH=75% after one month (T1) and three months (T2). Samples were analyzed using a HPLC method as described in Example2.

C. Conclusion

The data at both 25° C. and at 40° C. have shown that the blending process inhibits formation of amounts of the impurity RRT=1.22 to below the detection limit of the HPLC method of Example 2.

Pharmaceutical compositions comprising ceftolozane and tazobactam with less than 0.15% (measured by HPLC according to Example 2) of the compound of formula (III) can be obtained as described herein.

TABLE 10
Excipients Used in Ceftolozane composition
Inactive
Concentration Ingredients
Amount, in Infusion Rationale for Database
Component Function mg/Vial Solution, % Inclusion (IID) Range
Citric acid Chelating  21 0.02 Used to prevent 0.0025 to 50%
agent discoloration and
degradation
Sodium Stabilizing 487 0.49 Used as a stabilizing  0.187 to 45%
Chloride agent agent for
ceftolozane sulfate
L-arginine Alkalizing 600i) 0.60 Used to adjust  0.29 to 88%
agent Q.S. for pH ceftolozane solution
adjustment pH
i)L-arginine is added as needed to achieve pH 6.5 ± 0.5; 600 mg per vial is considered a representative total amount.

TABLE 11
Unit Compositions of Ceftolozane/Tazobactam for Injection,
1000 mg/500 mg
Nominal
Composition
Component Function mg per Vial
Ceftolozane Ceftolozane Active 1147 
composition1) Sulfate
Citric Acid, Chelating  21
Anhydrous Agent
Sodium Stabilizing 487
Chloride Agent
L-Arginine Alkalizing   6002)
Agent Q.S. for pH
adjustment
Tazobactam Sodium3) Active 537
Nitrogen Processing Q.S.
Aidii)
Total Weight 2792 
1)Actual amount of ceftolozane composition will vary based on the measured potency. Ceftolozane sulfate, 1147 mg, corresponds to 1000 mg ceftolozane free base.
2)L-arginine is added as needed to achieve pH 6.5 ± 0.5; 600 mg per vial is considered a representative total amount.
3)Actual weight of tazobactam sodium will vary based on the measured potency. Tazobactam sodium 537 mg, corresponds to 500 mg tazobactam free acid
4) Nitrogen blanket is applied after powders are dispensed to the vial and prior to insertion of stopper.

A first aqueous solution comprising the ingredients in the Ceftolozane composition in Table 11 is lyophilized in the absence of tazobactam to provide the lyophilized Ceftolozane composition. The first aqueous solution comprises ceftolozane sulfate and the specific excipients in the preferred compositions, in an amount per unit dosage form provided by the quantities and functions as provided in Table 10. All excipients are compendial and typical for sterile pharmaceutical dosage forms, requiring no additional treatment prior to use in the formulation. The excipients are used in levels within the range established in other FDA approved products as described in the Inactive Ingredients Database (IID). A second solution comprising tazobactam acid and sodium bicarbonate is lyophilized in the absence of ceftolozane to obtain the Tazobactam Sodium Composition in Table 11. Subsequently, the lyophilized Tazobactam Sodium Composition is dry blended with the lyophilized Ceftolozane composition comprising tazobactam sodium and ceftolozane sulfate in a weight ratio providing 500 mg of tazobactam acid equivalent per 1,000 mg of ceftolozane active equivalent.

Jiang, Chun, Terracciano, Joseph, Damour, Nicole Miller, Fogliato, Giovanni, Donadelli, Giuseppe Alessandro, Resemini, Dario

Patent Priority Assignee Title
10420841, Mar 15 2013 Merck Sharp & Dohme LLC Ceftolozane antibiotic compositions
11278622, Mar 15 2013 Merck Sharp & Dohme LLC Ceftolozane antibiotic compositions
11452291, May 14 2007 The Research Foundation for The State University Induction of a physiological dispersion response in bacterial cells in a biofilm
11541105, Jun 01 2018 The Research Foundation for The State University of New York Compositions and methods for disrupting biofilm formation and maintenance
9320740, Mar 15 2013 Merck Sharp & Dohme LLC Ceftolozane-tazobactam pharmaceutical compositions
9872906, Mar 15 2013 Merck Sharp & Dohme LLC Ceftolozane antibiotic compositions
9925196, Mar 15 2013 Merck Sharp & Dohme LLC Ceftolozane-tazobactam pharmaceutical compositions
Patent Priority Assignee Title
4200575, Aug 06 1977 Takeda Chemical Industries, Ltd. 7[(2-Thiazolyl)-2-(oxoimino)acetamido]cephalosporin derivatives
4246405, Jun 03 1977 Fujisawa Pharmaceutical Company, Limited Method for preparation of β-lactam compound
4255431, Apr 14 1978 AstraZeneca AB Gastric acid secretion inhibiting substituted 2-(2-benzimidazolyl)-pyridines, pharmaceutical preparations containing same, and method for inhibiting gastric acid secretion
4264597, Jun 06 1978 FUJISAWA PHARMACEUTICAL CO , LTD , NO 3, 4-CHOME, DOSHOMACHI, HIGASHI-KU, OSAKA, JAPAN Cephalosporin analogues and processes for the preparation thereof
4267176, Nov 14 1977 FUJISAWA PHARMACEUTICAL CO , LTD Amino or acylamino-substituted pyrimidinyl cephalosporanic acid derivatives and pharmaceutical compositions thereof
4268509, Jul 10 1978 Fujisawa Pharmaceutical Co., Ltd. New cephem compounds and processes for preparation thereof
4284631, Jul 31 1978 Fujisawa Pharmaceutical Co., Ltd. 7-Substituted cephem compounds and pharmaceutical antibacterial compositions containing them
4291031, Feb 19 1979 Fujisawa Pharmaceutical Co., Ltd. 3-Phosphonocephalosporanic acid derivatives, and pharmaceutical composition comprising the same
4298529, Sep 12 1978 FUJISAWA PHARMACEUTICAL CO , LTD Alkoxyimino dioxy butyric acid derivatives
4299829, Jun 28 1976 Northern Telecom Limited 2-Lower alkyl-7-substituted-2 or 3-cephem 4-carboxylic acid compounds
4305937, Aug 17 1978 Fujisawa Pharmaceutical Co., Ltd. 2-Lower alkyl-7-substituted-2 or 3-cephem-4-carboxylic acid compounds and antibacterial pharmaceutical compositions containing them
4327093, Oct 24 1978 Fujisawa Pharmaceutical Co., Ltd. 3,7-Disubstituted-2 or 3-cephem-4-carboxylic acid compounds
4331665, Dec 29 1978 Fujisawa Pharmaceutical Co., Ltd. Cephem and cepham compounds
4332798, Dec 29 1978 Fujisawa Pharmaceutical Co., Ltd. 7-Amino-thia-diazole oxyimino derivatives of cephem and cephem compounds
4332800, Oct 12 1979 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4336253, Mar 11 1981 Eli Lilly and Company Cephalosporin antibiotics
4338313, Oct 12 1979 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4339449, Mar 27 1979 Fujisawa Pharmaceutical Company, Limited Analogous compounds of cephalosporins, and pharmaceutical composition comprising the same
4363807, Oct 17 1978 Fujisawa Pharmaceutical Company, Limited Cepham compounds
4367228, Oct 29 1980 Fujisawa Pharmaceutical Co., Ltd. Cephem compound and composition
4368325, Mar 06 1980 Fujisawa Pharmaceutical Co., Ltd. Processes for preparing 3-cephem compounds
4369312, Jul 04 1980 Fujisawa Pharmaceutical Co., Ltd. Method of producing oxo-containing azetidinone compounds
4370326, Sep 13 1977 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and composition
4381299, Dec 28 1979 Fujisawa Pharmaceutical Co., Ltd. 7-Amino-thiadiazole oxyimino derivatives of cephem and cepham compounds
4390534, Dec 29 1978 Fujisawa Pharmaceutical Co., Ltd. Cephem and cepham compounds
4394384, Sep 03 1979 Fujisawa Pharmaceutical Co., Ltd. Aminothiazole cycloalkenyloxyimino acetamido cephem derivatives
4402955, Oct 02 1981 Eli Lilly and Company Dioximino cephalosporin antibiotics
4405617, Feb 11 1980 Fujisawa Pharmaceutical Co., Ltd. 3-(Propynyltetrazol)thiomethyl-3-cephems
4407798, Aug 29 1980 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4409214, Nov 19 1979 Fujisawa Pharmaceutical, Co., Ltd. 7-Acylamino-3-vinylcephalosporanic acid derivatives and processes for the preparation thereof
4409215, Nov 19 1979 Fujisawa Pharmaceutical Co., Ltd. 7-Acylamino-3-substituted cephalosporanic acid derivatives and processes for the preparation thereof
4409217, Mar 14 1978 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4416879, Sep 08 1980 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4418058, Jun 23 1980 Shionogi & Co., Ltd. Protection of lyophilized betalactams from color formation
4420477, Nov 30 1979 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4423213, Nov 19 1979 Fujisawa Pharmaceutical Co., Ltd. 7-Acylamino-3-vinylcephalosporanic acid derivatives and processes for the preparation thereof
4425340, Dec 29 1978 FUJISAWA PHARMACEUTICAL CO LTD Cephem and cepham compounds
4425341, Mar 14 1977 Fujisawa Pharmaceutical Company, Limited Cephem compounds
4427677, Dec 31 1980 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4430499, Sep 08 1981 Eli Lilly and Company 7-[2-(2-Aminooxazol-4-yl)-2-(oximino)acetamido]cephalosporin antibiotics
4431642, Dec 01 1980 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4436912, Sep 08 1981 Eli Lilly and Company 7-[2-(2-Aminooxazol-4-yl)-2-(oximino)acetamido cephalosporin antibiotics and intermediates therefor
4438113, Dec 31 1980 Fujisawa Pharmaceutical Co., Ltd. 7-Acylaminocephalosporanic acid derivatives
4443443, Dec 17 1979 FUJISAWA PHARMACEUTICAL CO , LTD Cephem compounds
4443444, Aug 11 1980 FUJISAWA PHARMACEUTICAL CO , LTD Cephem compounds
4447429, Jul 10 1978 Fujisawa Pharmaceutical Co., Ltd. 7-[2-Alkoxyimino-2-(5-amino-1,2,4-thiadiazol-3-yl)acetamido]-3-cephems
4450270, Oct 02 1981 Eli Lilly and Company Dioximino cephalosporin antibiotics
4452851, Feb 02 1981 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4457928, Dec 01 1980 FUJISAWA PHARMACEUTICAL CO , LTD Cephem compounds
4462999, Nov 19 1979 Fujisawa Pharmaceutical Co., Ltd. 7-Acylamino-3-substituted cephalosporanic acid derivatives and processes for the preparation thereof
4463000, Dec 01 1980 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4463002, Mar 14 1978 FUJISAWA PHARMACEUTICAL CO , LTD NO 3,4-CHOME DOSHOMACHI, HIGASHI-KU OSAKA JAPAN Syn-isomers of 7-substituted-3-cephem-4-carboxylic acid esters
4463003, Dec 22 1982 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4464369, Mar 14 1977 Fujisawa Pharmaceutical Co., Ltd. 7-Acylamino-3-cephem-4-carboxylic acid derivatives and pharmaceutical compositions
4470980, Mar 07 1980 INTERx Research Corp. Method of increasing oral absorption of β-lactam antibiotics
4474779, Apr 09 1981 Yamanouchi Pharmaceutical Co., Ltd. Cephalosporin compounds
4477447, Dec 15 1980 Fujisawa Pharmaceutical Company, Limited 7-Acylaminocephalosporanic acid derivatives
4487768,
4495182, Jun 22 1981 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4496562, Mar 14 1978 Fujisawa Pharmaceutical Co., Ltd. 7-Substituted-3-cephem-4-carboxylic acid esters
4499088, Jan 04 1983 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4501739, Sep 08 1981 Eli Lilly and Company Thieno and furopyridinium-substituted cephalosporins
4515788, Aug 03 1981 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4521413, Sep 14 1981 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4529592, Jun 21 1982 Taiho Pharmaceutical Company, Limited Penicillin derivatives
4546101, Sep 10 1982 Fujisawa Pharmaceutical Co., Ltd. New cephem compounds useful for treating infectious diseases in human being and animals and processes for preparation thereof
4550102, Sep 14 1981 Fujisawa Pharmaceutical Co. Ltd. Cephem compounds
4559334, Nov 10 1980 Astellas Pharma INC 7-Substituted-3-vinyl-3-cephem compounds and processes for production of the same
4562073, Dec 24 1982 Taiho Pharmaceutical Company Limited Penicillin derivatives
4563449, Jul 19 1982 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4577014, Sep 08 1981 Eli Lilly and Company Thieno and furopyridinium-substituted cephalosporins
4584290, Dec 06 1982 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4585872, Jul 10 1978 Fujisawa Pharmaceutical Co., Ltd. Cephem intermediates
4590186, Jan 16 1984 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds having 3-bicyclic heterocyclic cation groups
4600772, May 26 1978 Glaxo Group Limited Cephalosporin antibiotics
4608373, Dec 13 1982 YAMANOUCHI PHARMACEUTICAL CO , LTD Cephem compounds
4609730, Nov 22 1982 Fujisawa Pharmaceutical Co., Ltd. 7-[substituted imino-2-(2-aminothiazol-4-yl)-acetamido]-3(2,2-dihalovinyl or ethynyl)-3-cephem-4-carboxylic acid (syn isomers), having antimicrobial activities
4616083, Aug 22 1983 Shionogi & Co., Ltd. Stable antibacterial lyophilizates
4622318, Oct 31 1983 Fujisawa Pharmaceutical Co., Ltd. 3-(substituted-ethenyl or ethynyl-thiomethyl) cephems
4626384, Apr 06 1984 Taiho Pharmaceutical Company, Limited Penam derivatives
4631274, Mar 09 1984 Fujisawa Pharmceutical Co., Ltd. Halovinyl cephem compounds
4640915, Mar 29 1982 Fujisawa Pharmaceutical Co., Ltd. 1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid derivatives
4647556, Mar 04 1982 Hoechst Aktiengesellschaft Cephalosporin derivatives
4667028, May 07 1983 Hoechst Aktiengesellschaft Process for the preparation of cephem compounds
4690921, Oct 11 1983 Yamanouchi Pharmaceutical Co., Ltd. Cephalosporin compounds and salts thereof
4692443, Oct 17 1983 Eli Lilly and Company 3-bicyclicpyridinium-methyl cephalosporins
4698337, Nov 13 1978 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and processes for preparation thereof
4699980, Feb 10 1984 Yamanouchi Pharmacutical Co., Ltd. Process for preparing 3-substituted-methyl-3-cephem derivatives
4703046, Sep 08 1978 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and processes for preparation thereof
4705851, Sep 28 1984 Fujisawa Pharmaceutical Co., Ltd. Process for the preparation of 3-phosphoniummethyl-3-cephem compounds
4735937, Oct 27 1970 FUJISAWA PHARMACEUTICAL CO , LTD 8-oxo-5-thia-1-azabicyclo(4,2,0)oct-2-ene compounds
4748172, Dec 10 1984 Eli Lilly and Company 3-bicyclicpyridinium-methyl cephalosporins
4761410, Jan 14 1985 Fujisawa Pharmaceutical Co., Ltd. Cephem Compounds
4764606, Aug 01 1985 Yamanouchi Pharmaceutical Co., Ltd. 7-formylaminocephalosporin compounds
4769183, Mar 09 1983 NIPPON SHINYAKU CO , LTD Stabilization of azulene derivatives
4808617, Dec 18 1985 Bristol-Myers Company Lyophilized or precipitated cephalosporin zwitterion and salt combination
4808711, Jan 21 1985 Sankei Pharmaceutical Co., Ltd.; Nippon Pharmaceutical Development Institute Co. Cephalosporin derivatives
4822785, Jul 10 1986 Eisai Co., Ltd. Cephalosporin injection
4822787, Nov 24 1986 Fujisawa Pharmaceutical Co., Ltd. 3-pyrrolidinylthio-1-azabicyclo(3.2.0)-hept-2-ene-2-carboxylic acid compounds and antimicrobial activity thereof
4833134, Aug 19 1986 Takeda Chemical Industries, Ltd. Cephem compounds
4861769, Feb 18 1985 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4868174, Mar 17 1986 Fujisawa Pharmaceutical Co., Ltd. 3,7-Disubstituted-3-cephem compounds
4871730, Jul 18 1974 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4882434, Oct 29 1986 Takeda Chemical Industries, Ltd. Gamma-lactonecarboxylic acid derivatives and their use as antibacterial agents or intermediates
4921852, Nov 24 1986 FUJISAWA PHARMACEUTICAL CO , LTD 3-pyrrolidinylthio-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid compounds
4923857, Feb 22 1988 Fujisawa Pharmaceutical Company, Ltd 3-Alkenyl-1-azabicyclo(3.2.0)hept-2-ene-2-carboxylic acid compounds
4925934, Mar 02 1988 OTSUKA CHEMICAL CO , LTD Process for preparation of beta-lactam derivatives
4927818, Sep 22 1986 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and processes for preparation thereof
4935507, Aug 19 1987 Astellas Pharma INC Crystalline 7-(2-(2-aminothiazol-4-yl)-2-hydroxyiminoacetamido)-3-vinyl-3-cephem-4-c arboxylic acid (syn isomer)
4943567, May 30 1987 KYOTO PHARMACEUTICAL INDUSTRIES, LTD Cephalosporin compound and pharmaceutical composition thereof
4952578, Sep 14 1987 Fujisawa Pharmaceutical Co., Ltd. Cephem compound and a process for preparation thereof
4960766, Nov 22 1985 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
4963543, Nov 24 1986 Fujisawa Pharmaceutical Co., Ltd. 3-pyrolidinylthio-1-azabicyclo-[3.2.0]hept-2-ene-2-carboxylic acid derivatives and processes for the preparation thereof
4963544, May 23 1988 Fujisawa Pharmaceutical Company, Ltd. 3-pyrrolidinylthio-1-azabicyclo[3.2.0]-hept-2-ene-2-carboxylic acid compounds
4971962, May 11 1988 Lucky, Ltd. Cephalosporin compounds
4982596, Jan 26 1990 Illinois Tool Works Inc Die for manufacturing a fastener blank
5036064, Aug 14 1987 F HOFFMANN-LA ROCHE & CO AKTIENGESELLSCHAFT; Hoffmann-La Roche Inc Cephalosporins with sulfur-containing oxyimino side chain
5071979, May 12 1981 Sanofi-Aventis Deutschland GmbH Cephalosporin derivatives
5073550, Aug 09 1988 Hoffmann-La Roche Inc. Cephalosphorins with sulfur-containing oxyimino side chain
5081116, Apr 12 1989 Yamanouchi Pharmaceutical Co., Ltd. Cephalosporin derivatives
5095012, Aug 23 1990 Bristol-Myers Squibb Company Antibiotic C-7 catechol-substituted cephalosporin compounds, compositions, and method of use thereof
5102877, Apr 28 1989 Fujisawa Pharmaceutical Co., Ltd. 1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid compounds
5104866, Aug 10 1987 Fujisawa Pharmaceutical Company, Ltd. Water-soluble antibiotic composition and water-soluble salts of cephem compounds
5108997, Sep 22 1986 Fujisawa Pharmaceuticals Co., Ltd. Cephem compounds
5109130, Feb 27 1990 Fujisawa Pharmaceutical Co., Ltd. Cephem compound
5138066, Aug 09 1988 Hoffmann-La Roche, Inc. Intermediates for cephalosporins with sulfur-containing oxyimino side chain
5159070, Jul 26 1979 AVENTIS PHARMA S A Process for the preparation of derivative of 7-[(2-hydroxyimino)-acetamido]-cephalosporanic acid
5162520, Sep 22 1986 Fujisawa Pharmaceutical Co., Ltd. Intermediates for cephem compounds
5173485, Mar 09 1988 Fujisawa Pharmaceutical Company, Ltd. Cephem compounds
5179485, Jan 11 1989 Sony Corporation Optical encoder for disc apparatus
5187160, Mar 08 1989 Fujisawa Pharmaceutical Co., Ltd. Cephem compound
5194432, Nov 22 1985 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
5210080, Sep 07 1987 FUJISAWA PHARMACEUTICAL CO , LTD Cephem compounds
5215982, Nov 10 1989 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
5215983, Oct 23 1989 Fujisawa Pharmaceutical Co., Ltd. Carbapenem compounds
5219848, Aug 11 1989 ICI Pharma; Imperial Chemical Industries PLC Cephem compounds
5234920, Aug 23 1990 Bristol-Myers Squibb Company Antibiotic C-7 catechol-substituted cephalosporin compounds, compositions, and method of use thereof
5244890, Jun 06 1988 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
5281589, Jun 15 1991 Cheil Foods & Chemicals, Inc. 3-fused pyridiniummethyl cephalosporins
5286721, Oct 15 1990 Fujisawa Pharmaceutical Co., Ltd. 1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid compounds
5319140, Aug 09 1988 Hoffmann-La Roche Inc. Intermediates for cephalosporins with sulfur containing oxyimino side chain
5329002, Mar 31 1988 Hoffmann-La Roche Inc. Antibacterial cephalosporin compounds
5336768, May 24 1988 Hoffmann-La Roche Inc. Antibacterial cephalosporin compounds
5366970, Mar 09 1988 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and pharmaceutically acceptable salts thereof
5389627, Jun 05 1993 Cheil Foods & Chemicals Cephem compounds
5498787, Apr 20 1994 Standard Chemical & Pharmaceutical Co., Ltd. Method for preparing cephalosporin derivatives
5523400, Apr 16 1993 HOFFMAN-LAROCHE INC Cephalosporin antibiotics
5637580, Oct 24 1988 The Procter & Gamble Company Antimicrobial penem-quinolones
5646139, Oct 24 1988 The Procter & Gamble Company Antimicrobial carbapenem quinolones
5648346, Oct 24 1988 The Procter & Gamble Company Antimicrobial carbacephem-quinolones
5656623, Oct 24 1988 The Procter & Gamble Company Antimicrobial lactam-quinolones
5661144, Aug 16 1994 Meiji Seika Kabushiki Kaisha Cephem derivatives with 3-substituted bis heterocycles
5663163, Sep 07 1987 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and processes for preparation thereof
5763603, Nov 06 1993 Taiho Pharmaceutical Co., Ltd. Crystalline tazobactam, and its production and use
6207661, Feb 22 1999 Baxter International Inc.; Baxter International Inc Premixed formulation of piperacillin sodium and tazobactam sodium injection
6214818, Apr 04 1996 Shionogi & Co., Ltd. Cephem compounds and pharmaceutical compositions containing the same
6458950, Apr 04 1997 Shionogi & Co., Ltd. Compounds and pharmaceutical compositions containing the same
6518263, Nov 27 1998 Shionogi & Co., Ltd. Imidazo[4,5-b]pyridiniummethyl-containing cephem compounds having broad antibacterial spectrum
6660855, Aug 11 2000 OTSUKA CHEMICAL CO , LTD Crystals of penicillin and process for the production thereof
6774104, Jul 01 1999 Astellas Pharma INC Stabilized pharmaceutical composition in lyophilized form
6800621, Nov 27 1998 Shionogi & Co., Ltd. Imidazo[4,5-b]-pyridiniummethyl-containing cephem compounds having broad antibacterial spectrum activity
6878686, May 24 2002 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
6936711, Jun 07 2002 Orchid Chemicals and Pharmaceuticals Limited Process for preparation of penam derivatives from cepham derivatives
6974797, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
6995138, May 24 2002 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7067481, May 23 2003 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7067482, Jul 11 2003 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7112565, Jul 01 1999 Astellas Pharma INC Stabilized pharmaceutical composition in lyophilized form
7129232, Oct 30 2002 Astellas Pharma INC Cephem compounds
7179801, May 01 2001 Astellas Pharma INC Cephem compounds
7192943, Sep 18 2003 Astellas Pharma INC Cephem compounds
7273935, Aug 21 2003 ORCHID CHEMICALS & PHARMACEUTICALS LTD Process for the preparation of 3-methylcepham derivatives
7279458, Jul 11 2003 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7304075, Apr 23 2001 DAIICHI PHARMACEUTICAL CO , LTD Stabilized liquid preparation
7332471, May 23 2003 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7341993, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide—cephalosporin antibiotics
7384928, Mar 18 2002 SHIONOGI & CO , LTD Broad spectrum cephem compounds
7417143, Apr 07 2004 ORCHID CHEMICALS & PHARMACEUTICALS LTD Process for the preparation of Tazobactam in pure form
7547777, Oct 10 2003 OTSUKA CHEMICAL CO , LTD ; TAIHO PHARMACEUTICAL CO , LTD Penam crystals and process for producing the same
7553962, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7601690, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7612037, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7649080, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7655621, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7674898, May 01 2001 OTSUKA CHEMICAL CO , LTD ; TAIHO PHARMACEUTICAL CO , LTD Anhydrous crystal of β-lactam compound and method for preparation thereof
7728127, Oct 12 2001 Theravance Biopharma Antibiotics IP, LLC Cross-linked glycopeptide-cephalosporin antibiotics
7842683, Aug 16 1999 Revaax Pharmaceuticals, LLC Neurotherapeutic compositions and method
7915229, Apr 07 2004 Baxter Healthcare Corporation Compositions containing piperacillin and tazobactam useful for injection
8133883, Apr 14 2003 Baxter Healthcare Corporation Compositions containing piperacillin and tazobactam useful for injection
8476425, Sep 27 2012 Merck Sharp & Dohme LLC Tazobactam arginine compositions
20020115650,
20020193587,
20030130173,
20030232983,
20040248875,
20050004094,
20050096306,
20050171077,
20050228176,
20060051412,
20060084639,
20060099253,
20060173177,
20060241017,
20060269485,
20060287244,
20060293516,
20070054899,
20070116770,
20070219191,
20070286817,
20070286818,
20080015156,
20080160067,
20080233196,
20090098088,
20090137460,
20090155387,
20090156517,
20090156518,
20090186865,
20090227554,
20090274662,
20090275552,
20090291102,
20090311234,
20100040548,
20100286031,
20110044917,
20110136763,
20110190252,
20110257079,
20130065874,
20140187528,
20140206659,
20140213567,
20140262868,
20140274989,
20140274990,
20140274991,
20140274992,
20140274993,
20140274994,
20140274995,
20140274996,
20140274997,
20140274998,
20140275000,
20140303136,
20140309205,
AU2002952355,
AU2003904813,
AU2003905084,
AU614793,
AU707730,
CA1235689,
CA2140701,
CN200410067367,
CN200810092568,
CN200810238479,
CN200910169647,
CN201010557481,
CN201110061045,
CN99100092,
EP47977,
EP97446,
EP111934,
EP137440,
EP137442,
EP138552,
EP318767,
EP664117,
EP711774,
EP771803,
EP1134222,
EP1154770,
EP1273586,
EP1285923,
EP1468697,
EP1554287,
EP1671974,
EP1686131,
EP1759697,
EP1787641,
EP1959933,
EP1974721,
EP2015755,
EP2062581,
EP2062582,
EP2062585,
EP2136844,
EP2305251,
JP2005162670,
JP2088582,
JP2117678,
JP4288086A,
JP5222058,
JP6056848,
JP6128268,
JP62103092,
JP62158290,
JP63051388,
JP63051389,
RE33778, Jul 28 1977 Astellas Pharma INC 1,3-dithietane-2-carboxylic acid penicillin and cephalosporin derivatives
WO50035,
WO2090363,
WO2090364,
WO2092605,
WO2102378,
WO3066053,
WO3104241,
WO2004019901,
WO2004039776,
WO2004066976,
WO2004098643,
WO2005074925,
WO2006044600,
WO2006045006,
WO2006088305,
WO2007065862,
WO2007086011,
WO2007086013,
WO2007086014,
WO2007099396,
WO2007129176,
WO2007145866,
WO2007145868,
WO2008030469,
WO2008065247,
WO2008075207,
WO2008101743,
WO2008113177,
WO2009048603,
WO2009122252,
WO2009134948,
WO2010014285,
WO2010142241,
WO2011101710,
WO2011112435,
WO2011127200,
WO9512601,
WO9741128,
WO4915,
WO3078440,
WO2004048551,
WO2005005436,
WO2009105782,
WO2013036783,
WO2014052799,
WO9928308,
WO9964049,
/////////
Executed onAssignorAssigneeConveyanceFrameReelDoc
May 22 2014CALIXA THERAPEUTICS, INC.(assignment on the face of the patent)
Jun 03 2014CUBIST PHARMACEUTICALS, INC CALIXA THERAPEUTICS, INC ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0339960017 pdf
Aug 04 2014JIANG, CHUNCALIXA THERAPEUTICS, INC ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0339960017 pdf
Sep 15 2014FOGLIATO, GIOVANNIACS DOBFAR S P A ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0339960001 pdf
Sep 15 2014DONADELLI, GIUSEPPE ALESSANDROACS DOBFAR S P A ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0339960001 pdf
Sep 15 2014ACS DOBFAR S P A CALIXA THERAPEUTICS, INC ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0339960017 pdf
Sep 18 2014RESEMINI, DARIOACS DOBFAR S P A ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0339960001 pdf
Jun 10 2015CALIXA THERAPEUTICS, INC Merck Sharp & Dohme CorpASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0371980658 pdf
Apr 07 2022Merck Sharp & Dohme CorpMerck Sharp & Dohme LLCMERGER SEE DOCUMENT FOR DETAILS 0611020145 pdf
Date Maintenance Fee Events
Aug 21 2018M1551: Payment of Maintenance Fee, 4th Year, Large Entity.
Aug 10 2022M1552: Payment of Maintenance Fee, 8th Year, Large Entity.


Date Maintenance Schedule
Mar 03 20184 years fee payment window open
Sep 03 20186 months grace period start (w surcharge)
Mar 03 2019patent expiry (for year 4)
Mar 03 20212 years to revive unintentionally abandoned end. (for year 4)
Mar 03 20228 years fee payment window open
Sep 03 20226 months grace period start (w surcharge)
Mar 03 2023patent expiry (for year 8)
Mar 03 20252 years to revive unintentionally abandoned end. (for year 8)
Mar 03 202612 years fee payment window open
Sep 03 20266 months grace period start (w surcharge)
Mar 03 2027patent expiry (for year 12)
Mar 03 20292 years to revive unintentionally abandoned end. (for year 12)